Language selection

Search

Patent 2914770 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2914770
(54) English Title: DELAYED RELEASE CYSTEAMINE BEAD FORMULATION, AND METHODS OF MAKING AND USING SAME
(54) French Title: PREPARATION DE BILLES DE CYSTEAMINE A LIBERATION RETARDEE
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 9/52 (2006.01)
  • A61K 31/145 (2006.01)
  • A61P 3/00 (2006.01)
  • A61P 25/00 (2006.01)
(72) Inventors :
  • POWELL, KATHLENE (United States of America)
  • MUTTAVARAPU, RAMESH (United States of America)
(73) Owners :
  • HORIZON THERAPEUTICS U.S. HOLDING LLC (United States of America)
(71) Applicants :
  • RAPTOR PHARMACEUTICALS INC. (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued: 2016-09-27
(86) PCT Filing Date: 2014-06-17
(87) Open to Public Inspection: 2014-12-24
Examination requested: 2016-01-05
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2014/042607
(87) International Publication Number: WO2014/204881
(85) National Entry: 2015-12-07

(30) Application Priority Data:
Application No. Country/Territory Date
61/835,965 United States of America 2013-06-17

Abstracts

English Abstract

An enteric-coated bead dosage form of cysteamine, and related methods of manufacture and use, are disclosed.


French Abstract

L'invention concerne une forme pharmaceutique de billes de cystéamine gastro-résistantes, et des méthodes associées de fabrication et d'utilisation.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS:
1. A pharmaceutical dosage form comprising delayed-release cysteamine
beads, the
beads comprising:
(i) a core particle comprising a mixture of cysteamine bitartrate and a
binder, and
(ii) an enteric membrane surrounding the core particle;
wherein the beads have a distribution of particle sizes in a range of about
0.7 mm to about
2.8 mm;
wherein the enteric membrane begins to dissolve within a pH range of about 4.5
to about
6.5;
wherein the enteric membrane is present in an amount in a range of about 20%
to about 40%
by weight, based on the weight of the core particles; and
wherein the pharmaceutical dosage form, upon administration to cystinosis
patients at 425
mg to 1300 mg free cysteamine base during treatment, provides:
(a)a mean Cmax upon oral dosing, in a range of about 3.5 1.7 mg/L or in a
range
of 80% to 125% thereof; and
(b)a mean AUC (0-inf_D) upon oral dosing, in a range of about 1.08
0.46 min*mg/L/mg or in a range of 80% to 125% thereof.
2. The pharmaceutical dosage form of claim 1, wherein the cysteamine as
free base
comprises at least 10 wt.% of the core particle.
3. The pharmaceutical dosage form of claim 1, wherein the enteric membrane
is
present in an amount in a range of about 25% to about 35% by weight, based on
the
weight of the core particles.
4. The pharmaceutical dosage form of claim 1, wherein the cysteamine
bitartrate
comprises at least 50 wt.% of the core particle.
5. The pharmaceutical dosage form of claim 1, wherein the beads provide a
mean Cmax
and mean AUC (0-inf_D) upon oral dosing, fasted, when administered inside a
capsule shell
that are bioequivalent to the mean Cmax and mean AUC (0-inf_D) upon oral
dosing, fasted,
when administered without a capsule shell.
38

6. The pharmaceutical dosage form of claim 1, wherein the enteric membrane
comprises an enteric material that begins to dissolve at pH of about 5.5 in an
aqueous
solution.
7. The pharmaceutical dosage form of claim 1, wherein the pharmaceutical
dosage
form, upon administration to cystinosis patients at 425 mg to 1300 mg free
cysteamine
base during treatment, provides:
(a)a mean Cmax upon oral dosing, in a range of about 3.5 1.7 mg/L; and
(b)a mean AUC (0-inf_D) upon oral dosing, in a range of about 1.08 0.46
min*mg/L/mg.
8. The pharmaceutical dosage form of claim 1, wherein the pharmaceutical
dosage
form, upon administration to cystinosis patients at 425 mg to 1300 mg free
cysteamine
base during treatment, provides:
(a)a mean Cmax upon oral dosing, of 3.5 mg/L or in a range of 80% to 125%
thereof; and
(b)a mean AUC (0-inf_D) upon oral dosing, of 1.08 min*mg/L/mg or in a range of

80% to 125% thereof.
9. A pharmaceutical dosage form, comprising delayed-release cysteamine
beads, the beads
comprising:
(i) a core particle comprising cysteamine or a pharmaceutically acceptable
salt thereof
and a binder, and
(ii) an enteric membrane surrounding the core particle,
wherein the beads have a distribution of particle sizes in a range of about
0.7 mm to about 2.8
mm;
wherein the enteric membrane begins to dissolve within a pH range of about 4.5
to about 6.5;
wherein the enteric membrane is present in an amount in a range of about 20%
to about 40% by
weight, based on the weight of the core particles; and
wherein the pharmaceutical dosage form, upon administration in a capsule to
fasted healthy
normal subjects at 600 mg free cysteamine base, provides:
(a) a mean Cmax upon oral dosing in a range of 2.3. Ø6 mg/L or in a range of
80% to
125% thereof; and
39

(b) a mean AUC (0-inf_D) upon oral dosing in a range of 0.84. ~Ø19
min*mg/L/mg or
in a range of 80% to 125% thereof.
10. The pharmaceutical dosage form of claim 9, wherein the enteric membrane
is present in
an amount in a range of about 25% to about 35% by weight, based on the weight
of the core
particles.
11. The pharmaceutical dosage form of claim 9, wherein the particle sizes
of the beads are in
a range of about 0.7 mm to about 2.5 mm.
12. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by at least 80% by weight of the beads having a particle size in
a range of about
850 µm to about 1180 µm.
13. The pharmaceutical composition of claim 9, wherein 5% or less of the
beads by weight
are retained on a #12 mesh (1.68 mm) screen and 10% or less by weight pass
through a #20 mesh
(0.84 mm) screen.
14. The pharmaceutical composition of claim 9, wherein the distribution of
bead sizes is
characterized by less than 5% by weight of the beads being retained on a 1400
µm sieve.
15. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by less than 30% by weight of the beads being retained on a 1180
µm sieve.
16. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by less than 70% by weight of the beads being retained on a 1000
µm sieve.
17. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by less than 20% by weight of the beads being retained on a 850
µm sieve.
18. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by at least 15% by weight of the beads being retained on a 1180
µm sieve.

19. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by at least 50% by weight of the beads being retained on a 1000
µm sieve.
20. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by at least 10% by weight of the beads being retained on a 850
µm sieve.
21. The pharmaceutical dosage form of claim 9, wherein the distribution of
bead sizes is
characterized by a median particle size in a range of about 850 µm to about
1180 µm.
22. The pharmaceutical dosage form of claim 9, wherein the bead core
particle further
comprises a filler.
23. The pharmaceutical dosage form of claim 9, wherein the cysteamine as
free base is
present in the bead core particle in an amount of at least 10 wt. %.
24. The pharmaceutical dosage form of claim 9, wherein the cysteamine or
pharmaceutically
acceptable salt thereof is a pharmaceutically acceptable salt of cysteamine.
25. The pharmaceutical dosage form of claim 9, wherein 5% or less of the
bead core particles
by weight are retained on a #12 mesh (1.68 mm) screen and 10% or less by
weight pass through
a #20 mesh (0.84 mm) screen.
26. The pharmaceutical dosage form of claim 9, wherein the enteric-coated
beads are
characterized by acid resistance such that not more than 10% of the cysteamine
in the beads is
dissolved after a period of two hours in a 0.1N HCl solution.
27. The pharmaceutical dosage form of claim 9, wherein the enteric-coated
beads are
characterized by dissolution such that 80% of the cysteamine or
pharmaceutically acceptable salt
thereof is released within 20 minutes in a solution buffered at pH 6.8.
41

28. The pharmaceutical dosage form of claim 9, further comprising a capsule
shell enclosing
the plurality of beads.
29. The pharmaceutical dosage form of claim 9, wherein the beads provide a
mean Cmax and
mean AUC (0-inf_D) upon oral dosing, fasted, when administered inside a
capsule shell that are
bioequivalent to the mean Cmax and mean AUC (0-inf_D) upon oral dosing,
fasted, when
administered without a capsule shell.
30. The pharmaceutical dosage form of claim 9, wherein the enteric membrane
comprises an
enteric material that begins to dissolve at pH of about 5.5 in an aqueous
solution.
31. The pharmaceutical dosage form of claim 9, wherein the pharmaceutical
dosage form,
upon administration in a capsule to fasted healthy normal subjects at 600 mg
free cysteamine
base, provides:
(a) a mean Cmax upon oral dosing in a range of 2.3. ~Ø6 mg/L; and
(b) a mean AUC (0-inf_D) upon oral dosing in a range of 0.84. ~Ø19
min*mg/L/mg.
32. The pharmaceutical dosage form of claim 9, wherein the pharmaceutical
dosage form,
upon administration in a capsule to fasted healthy normal subjects at 600 mg
free cysteamine
base, provides:
(a) a mean Cmax upon oral dosing of 2.3 mg/L or in a range of 80% to 125%
thereof; and
(b) a mean AUC (0-inf_D) upon oral dosing of 0.84 min*mg/L/mg or in a range of
80%
to 125% thereof.
42

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02914770 2016-03-29
WO 2014/204881 PCT/US2014/042607
DELAYED RELEASE CYSTEA1VIINE BEAD FORMULATION, AND METHODS OF
MAKING AND USING SAME
[0001]
BACKGROUND
Field of the Disclosure
[0002] The disclosure relates generally to delayed release formulations of
cysteamine and
pharmaceutically acceptable salts thereof, and related methods of making and
treatment, e.g.
treatment of cystinosis and other metabolic and neurodegenerative diseases
including non-
alcoholic fatty liver disease (NAFLD), Huntingon' s disease, Parkinson' s
disease, Rett
Syndrome and others, use as free radical and radioprotectants, and as hepto-
protectant agents.
More particularly, the disclosure relates to enteric coated beads comprising
cysteamine or a
pharmaceutically acceptable salt thereof.
Brief Description of Related Technology
[0003] Cystinosis is a rare, autosomal recessive disease caused by intra-
lysosomal
accumulation of the amino acid cystine within various tissues, including the
spleen, liver,
lymph nodes, kidney, bone marrow, and eyes. Nephropathic cystinosis is
associated with
kidney failure that necessitates kidney transplantation. A specific treatment
for nephropathic
cystinosis is the sulfhydryl agent, cysteamine. Cysteamine has been shown to
lower
intracellular cystine levels, thereby reducing the rate of progression of
kidney failure in
children.
[0004] An enterically-coated cysteamine composition has been described, for
increasing
delivering of cysteamine to the small intestine and resulting in less frequent
dosing compared
to non enteric-coated cysteamine.
SUMMARY
[0005] One aspect of the disclosure provides a pharmaceutical dosage form
including a
plurality of cysteamine beads, the beads including a core particle including
cysteamine or a
pharmaceutically acceptable salt thereof and a pharmaceutically acceptable
excipient, and an
enteric membrane surrounding the core, wherein the plurality of beads is
characterized by a
distribution of particle sizes.
-1-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0006] Another aspect of the disclosure provides a pharmaceutical dosage form
including a
plurality of cysteamine beads, the beads including a core particle including
cysteamine or a
pharmaceutically acceptable salt thereof and a pharmaceutically acceptable
excipient, and an
enteric membrane surrounding the core, wherein the plurality of beads is
characterized by
irregular bead shapes.
[0007] Yet another aspect of the disclosure provides a pharmaceutical dosage
form
including a plurality of cysteamine beads, the beads including a core particle
including
cysteamine or a pharmaceutically acceptable salt thereof and a
pharmaceutically acceptable
excipient, and an enteric membrane surrounding the core, wherein the plurality
of beads is
characterized by a distribution of enteric membrane thicknesses.
[0008] Still another aspect of the disclosure provides a method of making a
pharmaceutical
dosage form, including any embodiment described herein, by a method including
coating a
core particle including cysteamine or a pharmaceutically acceptable salt
thereof and an
excipient with an enteric polymer to form an enteric membrane. The method can
include
sorting core particles prior to enteric coating, to provide a selected core
particle size
distribution. The method can also include sorting enteric coated beads to
provide a selected
bead size distribution.
[0009] Yet another aspect of the disclosure provides a method for treating a
patient in need
of cysteamine comprising administering to the patient a dosage form described
herein,
including any embodiment described herein.
[0010] Still another aspect of the disclosure provides dosage forms and
related methods
according the disclosure herein wherein the primary active component is
cystamine rather
than cysteamine or a pharmaceutically acceptable salt thereof.
[0011] For the compositions and methods described herein, optional features,
including but
not limited to components, compositional ranges thereof, substituents,
conditions, and steps,
are contemplated to be selected from the various aspects, embodiments, and
examples
provided herein.
[0012] Further aspects and advantages will be apparent to those of ordinary
skill in the art
from a review of the following detailed description. While the dosage form,
method of
making, and method of treatment are susceptible of embodiments in various
forms, the
description hereafter includes specific embodiments with the understanding
that the
disclosure is illustrative, and is not intended to limit the invention to the
specific
embodiments described herein.
-2-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
DETAILED DESCRIPTION
[0013] Described herein is pharmaceutical dosage form that includes a
plurality of
cysteamine beads, the beads including a core particle including cysteamine or
a
pharmaceutically acceptable salt thereof and a pharmaceutically acceptable
excipient, and an
enteric membrane surrounding the core particle. The plurality of beads can be
characterized
by a distribution of particle sizes. The plurality of beads can be
characterized by irregular
bead shapes. The plurality of beads can be characterized by a distribution of
enteric
membrane thicknesses. Also disclosed herein are a method for the preparation
of the dosage
form, including coating a core particle including cysteamine or a
pharmaceutically acceptable
salt thereof and an excipient with an enteric polymer to form the enteric
membrane.
Optionally, the core particle can be formed by a wet granulation method.
Optionally,
granules are sorted (e.g., via sieving) to a desired particle size range prior
to enteric coating,
and optionally again following enteric coating. Also disclosed herein are
treatment methods
including administering the dosage form to a patient in need thereof.
[0014] Cysteamine-containing, enteric-coated beads characterized by a
distribution of
particle sizes were shown to exhibit advantageous pharmacokinetics. Without
intending to be
bound by any particular theory, it is contemplated that the pharmacokinetics
are influenced
by the plurality of enteric-coated beads having a distribution of core
particle sizes.
[0015] Cysteamine-containing, enteric-coated beads characterized by a
irregular bead
shapes were shown to exhibit advantageous pharmacokinetics. Without intending
to be
bound by any particular theory, it is contemplated that the pharmacokinetics
are influenced
by the plurality of enteric-coated beads having irregular bead shapes.
[0016] Cysteamine-containing, enteric-coated beads characterized by a
distribution of
enteric membrane thicknesses were shown to exhibit advantageous
pharmacokinetics.
Without intending to be bound by any particular theory, it is contemplated
that the
pharmacokinetics are influenced by the plurality of enteric-coated beads
having a distribution
of enteric membrane thicknesses.
[0017] In one aspect the distribution of enteric membrane thicknesses can be
stated in
terms of weight gain of enteric membrane material based on the total weight of
the coated
beads. Thus, in one embodiment, the distribution of enteric membrane
thicknesses will be at
least 2% based on the total weight of the coated beads. In another embodiment,
the
distribution of enteric membrane thicknesses will be at least 3%. In another
embodiment, the
distribution of enteric membrane thicknesses will be at least 4%. In another
embodiment, the
-3-

CA 02914770 2015-12-07
WO 2014/204881
PCT/US2014/042607
distribution of enteric membrane thicknesses will be at least 5%. In another
embodiment, the
distribution of enteric membrane thicknesses will be at least 6%. In another
embodiment, the
distribution of enteric membrane thicknesses will be at least 7%. In another
embodiment, the
distribution of enteric membrane thicknesses will be at least 8% . In another
embodiment,
the distribution of enteric membrane thicknesses will be at least 9%. In
another embodiment,
the distribution of enteric membrane thicknesses will be at least 10%. In
another
embodiment, the distribution of enteric membrane thicknesses will be at least
11%. In
another embodiment, the distribution of enteric membrane thicknesses will be
at least 12%.
In another embodiment, the distribution of enteric membrane thicknesses will
be at least
13%. In another embodiment, the distribution of enteric membrane thicknesses
will be at
least 14%. For example, the difference in enteric membrane thickness from bead
to bead can
be in a range of +/- 1-7% based on the total weight of the coated beads. The
distribution of
enteric membrane thicknesses can be in a range of about 2% to about 14% based
on the
weight of the coated beads, or in a range of about 3% to about 13%, or in a
range of about 4%
to about 12%, or in a range of about 5% to about 11%, or in a range of about
6% to about
10%, or in a range of about 7% to 9%, or in a range of about 3% to 14%, or in
a range of
about 4% to 14%, or in a range of about 4% to 13%, or in a range of about 4%
to about 12%,
for example. In one embodiment, the absorption (AUC) of the dosage form when
dosed
orally is advantageously increased, compared to other dosage forms of
cysteamine. Without
intending to be bound by any particular theory, it is contemplated that the
increase in
absorption is influenced by the dosage form exhibiting a pseudo-extended
release profile.
The pseudo-extended release profile is contemplated to be influenced by one or
more factors,
including a distribution of enteric membrane thicknesses, a distribution of
bead particle sizes,
and the beads having irregular bead shapes. For example, in an embodiment
wherein the
beads have a distribution of enteric membrane thicknesses, it is contemplated
that for beads
which have a relatively thin coating, the coating will completely dissolve at
the trigger pH
relatively quickly to release the cysteamine composition, whereas for beads
having a
relatively thick coating the coating will take somewhat longer to completely
dissolve and
release the cysteamine composition. In another aspect, in an embodiment where
the beads
have a distribution of particle sizes and/or irregular bead shapes, it is
contemplated that the
gut transit time of the beads could be varied due to bead size and/or shape,
such that the
transit time until reaching the enteric membrane dissolution pH is varied,
thus contributing to
a pseudo-extended release profile. In another embodiment, the dosage form
exhibits
-4-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
substantially equivalent (e.g., bioequivalent) Cmax and/or AUC characteristics
when
administered orally inside a capsule shell or without a capsule shell.
[0018] The dosage form provides a progressive and predictable absorption
curve. In one
type of embodiment, the Tmax of the dosage form when dosed orally is
advantageously more
stable on a dose-to-dose basis, because the beads are individually enteric-
coated. A
predictable, consistent Tmax is highly advantageous for accomplishing a more
consistent,
sustained reduction of leukocyte cystine levels by use of cysteamine. For
example, process-
related variations in enteric membrane thickness or other influences on
enteric membrane
dissolution will affect only a fraction of the cysteamine in the dosage form
and will tend to
lead to the pseudo-extended release behavior described above. In contrast,
enteric-coated
capsules comprising cysteamine microspheres exhibited significant variability
in absorption
time from capsule to capsule.
[0019] In another embodiment, the dosage form exhibits advantageous storage
stability,
e.g. as measured by the amount of cystamine present following storage and/or
by the total
amount of related substances. The storage stability can be assessed following
storage at
typical ambient conditions (e.g. 25 C and 40% relative humidity) or at
accelerated stability
conditions involving increased temperature and/or humidity.
[0020] The dosage form and methods are contemplated to include embodiments
including
any combination of one or more of the additional optional elements, features,
and steps
further described below (including those shown in the figures and Examples),
unless stated
otherwise.
[0021] In jurisdictions that forbid the patenting of methods that are
practiced on the human
body, the meaning of "administering" of a composition to a human subject shall
be restricted
to prescribing a controlled substance that a human subject will self-
administer by any
technique (e.g., orally, inhalation, topical application, injection,
insertion, etc.). The broadest
reasonable interpretation that is consistent with laws or regulations defining
patentable
subject matter is intended. In jurisdictions that do not forbid the patenting
of methods that
are practiced on the human body, the "administering" of compositions includes
both methods
practiced on the human body and also the foregoing activities.
[0022] As used herein, the term "comprising" indicates the potential inclusion
of other
agents, elements, steps, or features, in addition to those specified.
[0023] As used herein, the term wt.% is the weight percent based on the total
weight, e.g.
of the core particle, or enteric membrane, or total bead, as described in
context. Unless stated
-5-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
otherwise, the wt.% is intended to describe the weight percent based on dry
weight (e.g., for a
core particle following drying).
[0024] All ranges set forth herein include all possible subsets of ranges and
any
combinations of such subset ranges. By default, ranges are inclusive of the
stated endpoints,
unless stated otherwise Where a range of values is provided, it is understood
that each
intervening value between the upper and lower limit of that range and any
other stated or
intervening value in that stated range, is encompassed within the disclosure.
The upper and
lower limits of these smaller ranges may independently be included in the
smaller ranges, and
are also encompassed within the disclosure, subject to any specifically
excluded limit in the
stated range. Where the stated range includes one or both of the limits,
ranges excluding
either or both of those included limits are also contemplated to be part of
the disclosure.
[0025] Unless expressly stated otherwise, all references to cysteamine herein
are intended
to encompass pharmaceutically-acceptable salts thereof, and for every
reference to
cysteamine herein the use of cysteamine bitartrate is specifically
contemplated as an
embodiment. As described in the Summary above, embodiments of the dosage forms
and
methods described herein can employ cystamine as the primary active component,
rather than
cysteamine or a pharmaceutically acceptable salt thereof.
[0026] Unless expressly stated otherwise, reference herein to a bead and
properties thereof
is intended to be interpreted as applying equally to a collection of beads
(e.g., a plurality of
such beads). Likewise, unless expressly stated otherwise, reference herein to
a core particle
and properties thereof is intended to be interpreted as applying equally to a
collection of core
particles (e.g., a plurality of such core particles).
[0027] As described above, a pharmaceutical dosage form is contemplated that
includes a
plurality of cysteamine beads, the beads including a core particle including
cysteamine or a
pharmaceutically acceptable salt thereof and a pharmaceutically acceptable
excipient, and an
enteric membrane surrounding the core particle, wherein the plurality of beads
is
characterized by a distribution of particle sizes.
[0028] In one embodiment, the particle sizes of the beads are in a range of
about 0.7 mm to
about 2.5 mm, or about 0.7 mm to about 2.8 mm, or about 0.8 mm to about 1.7
mm. For
example, the target bead size can be up to 2.5 mm with no more than 10 percent
variation
over this size, to a maximum size of 2.8 mm.
[0029] As the particle size of the beads becomes too small, the variability in
cysteamine
content increases. As the particle size becomes too large, the beads are too
large for use in
-6-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
drug products that are labeled to be administered via sprinkling (e.g., on
applesauce or other
soft foods, such as jellies) and swallowed without chewing, or administered
via an enteral
feeding tube. Also as the particle size increases, it was found that the
larger particles get
coated more than the smaller particles, resulting in lower relative assay when
compared to
use of smaller particles. To compensate, relatively more such beads would be
needed in
order to meet the label strength per capsule, but because salts such as
cysteamine bitartrate
already have a high molecular weight, filling a capsule shell with sufficient
large particles to
meet the label strength per capsule becomes difficult or impossible (e.g. to
fill a size 0
capsule to a 75mg strength of cysteamine free base). Accordingly the bead
particle size in
one type of embodiment is up to 1.7mm.
[0030] The distribution of bead particle sizes for various non-exclusive
embodiments of
the invention can be characterized in ways.
[0031] In one embodiment, the beads can be characterized by 5% or less of the
beads by
weight being retained on a #12 mesh (1.68mm) screen and 10% or less by weight
passing
through a #20 mesh (0.84mm) screen. In another embodiment, at least 80% by
weight of the
beads have a particle size in a range of about 850 lam to about 1180 lam, e.g.
as determined
by sieving.
[0032] The distribution of bead sizes can be characterized by a gradation test
via analytical
sieving. Thus, in another embodiment the distribution of bead sizes is
characterized by 0% of
the beads being retained on a 1700 lam sieve and less than 5% by weight of the
beads being
retained on a 1400 lam sieve. Optionally less than 30% by weight of the beads
are retained
on a 1180 lam sieve. Optionally less than 70% by weight of the beads are
retained on a 1000
lam sieve. Optionally less than 20% by weight of the beads are retained on a
850 lam sieve.
Optionally at least 15% by weight of the beads are retained on a 1180 lam
sieve. Optionally
at least 50% by weight of the beads are retained on a 1000 lam sieve.
Optionally at least 10%
by weight of the beads being retained on a 850 lam sieve.
[0033] Thus, for example, the distribution can be characterized by 0% of the
beads being
retained on a 1700 lam sieve and less than 5% by weight of the beads being
retained on a
1400 lam sieve, and about 20% to about 30% by weight of the beads being
retained on a 1180
lam sieve and then about 50% to about 70% (or about 55% to about 65%) by
weight of the
beads being retained on a 1000 lam sieve and then about 10% to about 20% by
weight of the
beads being retained on a 850 lam sieve.
-7-

CA 02914770 2016-03-29
WO 2014/204881 PCT/US2014/042607
[0034] In another embodiment, the distribution of bead sizes can be
characterized by a
median particle size in a range of about 850 p.m to about 1180 pm.
[0035] The bead core particle can comprise one or more excipients. In one type
of
embodiment, the excipients can include one or more fillers, binders, and
surfactants. Other
optional ingredients can include, but are not limited to, glidants,
lubricants, disintegrants,
swelling agents, and antioxidants.
[0036] Fillers include, but are not limited to, lactose, saccharose, glucose,
starch,
microcrystalline cellulose, microfine cellulose, mannitol, sorbitol, calcium
hydrogen
phosphate, aluminum silicate, amorphous silica, and sodium chloride, starch,
and dibasic
calcium phosphate dehydrate. In one type of embodiment, the filler is not
water soluble,
although it may absorb water. In one type of embodiment, the filler is a
spheronization aid.
Spheronization aids can include one or more of crospovidone, carrageenan,
chitosan, pectinic
acid, glycerides, 13-CD, cellulose derivatives, microcrystalline cellulose,
powdered cellulose,
polyplasdone crospovidone, and polyethylene oxide. In one embodiment, the
filler includes
microcrystalline cellulose.
[0037] Binders include, but are not limited to, cellulose ethers, methyl
cellulose, ethyl
cellulose, hydroxyethyl cellulose, propyl cellulose, hydroxypropyl cellulose,
lower-
substituted hydroxypropyl cellulose, hydroxypropylmethyl cellulose
(hypromellose, e.g.
hypromellose 2910, METHOCEL E), carboxymethyl cellulose, starch,
pregelatinized starch,
acacia, tragacanth, gelatine, polyvinyl pyrrolidone (povidone), cross-linked
polyvinyl
pyrrolidone, sodium alginate, microcrystalline cellulose, and lower-
substituted
hydroxypropyl cellulose. In one embodiment, the binders are selected from wet
binders. In
one type of embodiment, the binder is selected from cellulose ethers, e.g.
hypromellose.
[0038] Surfactants include, but are not limited to, anionic surfactants,
including sodium
lauryl sulfate, sodium deoxycholate, dioctyl sodium sulfosuccinate, and sodium
stearyl
fumarate, nonionic surfactants, including polyoxyethylene ethers, and
polysorbate 80, and
cationic surfactants, including quaternary ammonium compounds. In one
embodiment the
surfactant is selected from anionic surfactants, e.g. sodium lauryl sulfate.
[0039] Disintegrants include, but are not limited to, starch, sodium cross-
linked
carboxymethyl cellulose, carmellose sodium, carmellose calcium, cross-linked
polyvinyl
pyrrolidone, and sodium starch glycolate, low-substituted hydroxypropyl
cellulose,
hydroxypropyl starch.
-8-
*Trademark

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0040] Glidants include, but are not limited to, polyethylene glycols of
various molecular
weights, magnesium stearate, calcium stearate, calcium silicate, fumed silicon
dioxide,
magnesium carbonate, magnesium lauryl sulfate, aluminum stearate, stearic
acid, palmitic
acid, cetanol, stearol, and talc.
[0041] Lubricants include, but are not limited to, stearic acid, magnesium
stearate, calcium
stearate, aluminum stearate, and siliconized talc.
[0042] The amount of cysteamine free base in the core particle can be at least
10 wt.% or
at least 15 wt.%, or at least 20 wt.%, or at least 25 wt.% , or at least 30
wt.%. For example,
the amount of cysteamine bitartrate can be at least 50 wt.%, or at least 55
wt.%, or at least 60
wt.%, or at least 65 wt.%, or at least 70 wt.%, or at least 75 wt.%, or at
least 80 wt.%, or at
least 85 wt.% of the core particle, for example in a range of about 60 wt.% to
about 90 wt.%
or about 65 wt.% to about 85 wt.%. It is understood that any and all ranges
including these
values as endpoints is contemplated, for example, at least about 15 wt.% to
about 90 wt.%, or
at least about 20 wt.% to about 85 wt.%, or at least about 30 wt.% to about 85
wt.%, or at
least about 50 wt.% to about 90 wt.%. As the dose of cysteamine free base can
be up to
about 2 g/m2/day, and the amount of free base is relatively small compared to
the molecular
weight of salts (e.g. the bitartrate salt) it is preferred that the core
particle have as much active
ingredient as possible while allowing the creation and processing of core
particles.
[0043] The amount of filler in the core particle is not particularly limited.
In embodiments,
the amount of filler (e.g. microcrystalline cellulose) can be in a range of
about 10 wt.% to
about 30 wt.%, or about 16 wt.% to about 23 wt.%, or at least 19 wt.% or at
least 19.5 wt.%,
for example about 20 wt.%.
[0044] The amount of binder in the core particle is not particularly limited.
In
embodiments, the amount of binder (e.g. hypromellose) can be in a range of
about 1 wt.% to
about 10 wt.%, or about 2 wt.% to about 8 wt.%, or about 4 wt.% to about 6
wt.%, for
example about 5 wt.%.
[0045] The amount of surfactant, e.g. as a processing aid, in the core
particle is not
particularly limited. In embodiments, the amount of surfactant (e.g.
microcrystalline
cellulose) can be in a range of about 0.1 wt.% to about 1 wt.%, or about 0.2
wt.% to about 0.8
wt.%, or about 0.4 wt.% to about 0.6 wt.%, for example about 0.5 wt.%.
[0046] The enteric (gastro-resistant) membrane material, e.g. polymer, can be
one that will
dissolve in intestinal juices at a pH level higher than that of the stomach,
e.g. a pH of greater
than 4.5, such as within the small intestine, and therefore permit release of
the active
-9-

CA 02914770 2016-03-29
WO 2014/204881
PCT/US2014/042607
substance in the regions of the small intestine and substantially not in the
upper portion of the
GI tract. In one type of embodiment, the enteric material begins to dissolve
in an aqueous
solution at pH between about 4.5 to about 5.5. In another type of embodiment,
the enteric
material rapidly dissolves in an aqueous solution at pH between of about 5. In
another type
of embodiment, the enteric material rapidly dissolves in an aqueous solution
at pH between
of about 5.5.
[0047] For example, pH-sensitive materials will not undergo significant
dissolution until
the dosage form has emptied from the stomach. The pH of the small intestine
gradually
increases from about 4.5 to about 6.5 in the duodenal bulb to about 7.2 in the
distal portions
of the small intestine (ileum). In order to provide predictable dissolution
corresponding to
the small intestine transit time of about 3 hours (e.g., 2-3 hours) and permit
reproducible
release therein, the membrane should begin to dissolve within the pH range of
the duodenum,
and continue to dissolve at the pH range within the small intestine.
Therefore, the amount
(thickness) of enteric membrane should be sufficient to be substantially
dissolved during the
approximate three hour transit time within the small intestine (e.g., the
proximal and mid-
small intestine).
[0048] Enteric (gastro-resistant) materials can include, but are not limited
to, one or more
of the following: cross-linked polyvinyl pyrrolidone; non-cross linked
polyvinylpyrrolidone;
hydroxypropylmethyl cellulose phthalate, hydroxypropylmethyl cellulose acetate
succinate,
cellulose acetate succinate; cellulose acetate phthalate, hydroxypropylmethyl
cellulose
acetate succinate, cellulose acetate trimellitate; starch acetate phthalate;
polyvinyl acetate
phthalate; carboxymethyl cellulose; methyl cellulose phthalate; methyl
cellulose succinate;
methyl cellulose phthalate succinate; methyl cellulose phthalic acid half
ester; ethyl cellulose
succinate; carboxymethylamide; potassium methacrylatedivinylbenzene copolymer;

polyvinylalcohols; polyoxyethyleneglycols; polyethylene glycol; sodium
alginate;
galactomannone; carboxypolymethylene; sodium carboxymethyl starch; copolymers
of
acrylic acid and/or methacrylic acid with a monomer selected from the
following: methyl
methacrylate, ethyl methacrylate, ethyl acrylate, butyl methacrylate, hexyl
methacrylate,
decyl methacrylate, lauryl methacrylate, phenyl methacrylate, methyl acrylate,
isopropyl
acrylate, isobutyl acrylate, or octadecyl acrylate, e.g. EUDRAGIT -L and -S
series, including
L 100-55, L 30 D-55, L 100, S 100, L 12.5, and S 12.5, available from Evonik
Industries;
polyvinyl acetate; fats; oils; waxes; fatty alcohols; shellac; zein; gluten;
ethylacrylate-maleic
acid anhydride copolymer; maleic acid anhydride-vinyl methyl ether copolymer;
styrol-
maleic acid copolymer; 2-ethyl-hexyl-acrylate maleic acid anhydride; crotonic
acid-vinyl
-10-
*Trademark

CA 02914770 2016-03-29
WO 2014/204881 PCT/US2014/042607
acetate copolymer; glutaminic acid/glutamic acid ester copolymer;
carboxymethylethylcellulose glycerol monooctanoate; polyarginine;
poly(ethylene);
poly(propylene); poly(ethylene oxide); poly(ethylene terephthalate);
poly(vinyl isobutyl
ether); poly(vinyl chloride); and polyurethane. A combination of enteric
materials may also
be used. In one embodiment, the enteric material rapidly dissolves at pH 5.5
and higher, to
provide fast dissolution in the upper bowel. For example, the enteric material
can be selected
from a copolymer of methacrylic acid and methyl methacrylate, and a copolymer
of
methacrylic acid and ethyl acrylate. For example, an enteric polymer is
poly(methacrylic
acid co-ethyl acrylate)1:1 (EUDRAGIT L 30 D-55 and EUDRAGIT L100-55).
[0049] Examples of some enteric coatings are disclosed in U.S. Pat. No.
5,225,202,
including beeswax and glyceryl monostearate; beeswax, shellac and cellulose;
and cetyl
alcohol, mastic and shellac, as well as shellac and stearic acid (U.S. Pat.
No. 2,809,918);
polyvinyl acetate and ethyl cellulose (U.S. Pat. No. 3,835,221); and neutral
copolymer of
polymethacrylic acid esters (Eudragit L30D) (F. W. Goodhart et al., Pharm.
Tech., pp. 64-71
, April 1984); copolymers of methacrylic acid and methacrylic acid methylester
(Eudragits),
or a neutral copolymer of polymethacrylic acid esters containing metallic
stearates (Mehta et
al., U.S. Pat. Nos. 4,728,512 and 4,794,001). Such coatings comprise mixtures
of fats and
fatty acids, shellac and shellac derivatives and the cellulose acid phthlates,
e.g., those having
a free carboxyl content. See also Remington's Pharmaceutical Sciences, A.
Osol, ed., Mack
Pub. Co., Easton, Pa. (16th ed. 1980) at pages 1590-1593, and Z,eitova et al.
(U.S. Pat. No.
4,432,966), for descriptions of suitable enteric coating compositions.
[0050] One or more plasticizers can be added to enteric polymers in order to
increase their
pliability and reduce brittleness, as it is known in the art. Suitable
plasticizers are known in
the art and include, for example, butyl citrates, triethyl citrate, diethyl
phthalate, dibutyl
sebacate, PEGs (e.g. PEG 6000), acetyl triethyl citrate, and triacetin. In one
type of
embodiment, the plasticizer is triethyl citrate. While some enteric materials
are flexible and
do not require addition of plasticizers, more brittle polymers (e.g., Eudragit
US types,
Eudragit RL/RS, and Eudragit FS 30 D) benefit from plasticizers, e.g. in the
range of 5 wt.%
to 30 wt.% based on the dry polymer mass, e.g. about 8 wt.% to about 12 wt.%
triethyl citrate
with poly(methacrylic acid co-ethyl acrylate)1:1.
[0051] One or more anti-tacking agents (antiadherents) can also be added to an
enteric
coating mixture in order to reduce the tackiness of the film and prevent
agglomeration, as it is
known in the art. Anti-tacking agents include talc, and glyceryl monostearate,
fumed silica
(e.g., AEROSIL 200), precipitated silica (e.g., SWERNAT PQ), and magnesium
stearate, for
-11-
*Trademark

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
example. Anti-tacking agents can be used in any suitable quantity, for example
in a range of
about 10 wt.% to 100 wt.% based on dry polymer mass, or about 10 wt.% to about
50 wt.%,
or about 10 wt.% to about 30 wt. %, or about 15 wt.% to about 30 wt.%. For
example, in one
embodiment the amount of talc is in a range of 15 wt.% to about 30 wt.%, based
on dry
polymer mass.
[0052] One or more surfactants can also be added to an enteric coating mixture
in order to
improve substrate wettability and/or stabilize suspensions, as it is known in
the art.
Surfactants include Polysorbate 80, sorbitan monooleate, and sodium dodecyl
sulfate, for
example.
[0053] The enteric membrane can be formed by any suitable process. Coating
processes
include pan coating, fluid bed coating, and dry coating (e.g., heat dry
coating and electrostatic
dry coating), for example. Pan coating and fluid bed coating using solvent are
well
established processes. In liquid coating, the enteric material and optional
excipients (e.g.
pigments, plasticizers, anti-tacking agents) are mixed in an organic solvent
or water to form a
solution or dispersion. The coating solution or dispersion is sprayed into
solid dosage forms
in a pan coater or a fluid bed dryer and dried by hot air. For example, in a
Wurster fluid bed
coating process, the coating fluid is sprayed from the bottom of the fluid bed
apparatus,
whereas in an alternative the coating fluid is applied by top spraying, and in
another
alternative tangential spray is applied.
[0054] The amount of enteric material applied is sufficient to achieve desired
acid
resistance and release characteristics. For example, in one embodiment the
amount of enteric
membrane will be sufficient to meet United States Pharmacopeia (USP) <711>
requirements
(USP 36-NF 31) for delayed-release dosage forms, thereby not releasing 10.0
wt.% of drug
after 2 hours in 0.1N HC1. In another aspect, the formulation will be
sufficient to release at
least 80% of the active in 20 minutes in pH 6.8 buffer solution, e.g. using
the dissolution
method of USP 36-NF 31 section <711>.
[0055] In one type of embodiment, the enteric membrane is present in an amount
in a
range of about 20% to 40%, or 25% to about 35% as measured by the weight gain
compared
to the uncoated particle cores, or in a range of about 25% to about 31% weight
gain, or about
27% to about 31% weight gain, or about 28.5% to about 31% weight gain, based
on the
weight of the uncoated particle cores.
[0056] The beads with enteric membrane can be sorted (e.g., via sieving) to a
desired
particle size. In embodiments, the particle size range can be any particle
size range or
-12-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
combination thereof described above in connection with the core particles. In
one type of
embodiment, the particle size range will be the same as the particle size
range of the uncoated
core particles. For example, the beads can be sieved such that 5% or less of
the bead core
particles by weight are retained on a #12 mesh (1.68mm) screen and 10% or less
by weight
pass through a #20 mesh (0.84mm) screen.
[0057] Additional lubricant (glidant, anti-tack agent) can be added to the
coated beads in
powder form. Anti-tacking agents include talc, glyceryl monostearate, fumed
silica (e.g.,
AEROSIL 200), and precipitated silica (e.g., SIPERNAT PQ), for example. For
example talc
powder can be added to the coated beads, for example in an amount of 0.1 wt.%
to about 1
wt.% based on the total bead weight.
[0058] The formulation can include a capsule shell in which the beads are
disposed. Soft
and hard capsule shells are known. In one embodiment, the capsule shell is a
hard capsule
shell, e.g. a gelatin capsule shell or a vegetable-based hard capsule shell.
[0059] Thus, for example, one type of embodiment combining various of the
features
described above includes a pharmaceutical dosage form including a plurality of
cysteamine
beads, the beads including a core particle comprising cysteamine bitartrate, a
filler (optionally
microcrystalline cellulose), a binder (optionally hypromellose), and an
enteric membrane
(optionally Eudragit L30 D-55) surrounding the core, wherein the plurality of
beads is
characterized by a distribution of particle sizes in a range of about 0.7 mm
to about 2.5 mm,
wherein the enteric membrane is present in an amount in a range of about 20%
to about 40%
based on the weight of the bead core particles, and wherein the beads are
disposed in a
capsule shell.
[0060] Pharmacokinetics
[0061] As mentioned above, the dosage form can advantageously be designed have
one or
more pharmacokinetic characteristics, e.g. in humans.
[0062] In one embodiment, the pharmaceutical dosage form is characterized by a
mean
Tmax upon oral dosing, fasted, of greater than 75 minutes, or at least 110
minutes, or at least
2 hours, or at least 3 hours, or in a range of about 2.2 hours to about 3.48
hours, or about 2.22
hours to about 3.34 hours, or about 2.78 hours, or a Tmax in a range of 80% to
125%, or 80%
to 120% of such reference Tmax.
[0063] In another embodiment, the pharmaceutical dosage form is characterized
by a mean
Cmax upon oral dosing, fasted, in a range of about 22.16 [tmol/L to about
34.63 [tmol/L, or
-13-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
about 22.16 [tmol/L to about 33.24 [tmol/L, or about 22.7 [tmol /L, normalized
to a 450mg
dose, or a Cmax in a range of 80% to 125%, or 80% to 120% of such reference
Cmax. In
another embodiment, the pharmaceutical dosage form is characterized by a mean
Cmax_D
upon oral dosing in a range of about 0.004 to about 0.006 mg/L/mg.
[0064] In another embodiment, the pharmaceutical dosage form is characterized
by a mean
AUC (0-6 hours) upon oral dosing, fasted, in a range of about 60.74 p.mol=h/L
to about 94.91
p.mol=h/L, or about 60.74 p.mol=h/L to about 91.12 p.mol=h/L, or about 75.93
p.mol=h/L,
normalized to a 450 mg dose, or a bioequivalent AUC (0-6 hours) in a range of
80% to
125%, or 80% to 120% of such reference AUC (0-6 hours). In another embodiment,
the
pharmaceutical dosage form is characterized by a mean AUC (0-12 hours) upon
oral dosing
in a range of about 79.41 p.mol=h/L to about 124.08 p.mol=h/L, or about 79.41
p.mol=h/L to
about 119.11 p.mol=h/L, or about 99.26 p.mol=h/L, normalized to a 450 mg dose,
or a
bioequivalent AUC (0-12 hours) in a range of 80% to 125%, or 80% to 120% of
such
reference AUC (0-12 hours). In another embodiment, the pharmaceutical dosage
form is
characterized by a mean AUC (0-inf D) upon oral dosing in a range of about
0.86
min=mg/L/mg to about 1.35 min=mg/L/mg, or about 0.86 min=mg/L/mg to about 1.3
min=mg/L/mg, or a bioequivalent AUC (0-inf_D) in a range of 80% to 125%, or
80% to
120% of such reference AUC (0-inf D).
[0065] In example embodiments, any of the described pharmaceutical dosage
forms can be
characterized by providing mean pharmacokinetic parameters upon oral dosing,
fasted, of:
Tmax 183 90 minutes, Cmax 3.5 1.7 mg/L, and/or AUC (0-inf D) 1.08 0.46
min*mg/L/mg, or a bioequivalent Tmax, Cmax or AUC in a range of 80% to 125%,
or 80%
to 120% of such reference parameter.
[0066] In example embodiments, any of the described pharmaceutical dosage
forms can be
characterized by providing mean pharmacokinetic parameters upon oral dosing of
the whole
capsule, fasted, of: Tmax 194 38 minutes, Cmax 2.3 0 mg/L, and/or AUC (0-
inf D) 0.84
0.19 min*mg/L/mg, or a bioequivalent Tmax, Cmax or AUC in a range of 80% to
125%, or
80% to 120% of such reference parameter; and/or mean pharmacokinetic
parameters upon
oral dosing of the beads, sprinkled on applesauce, of: Tmax 190 61 minutes,
Cmax 2.3
0.7 mg/L, and/or AUC (0-inf D) 0.85 0.21 min*mg/L/mg, or a bioequivalent
Tmax, Cmax
or AUC in a range of 80% to 125%, or 80% to 120% of such reference parameter.
[0067] In another embodiment, the pharmaceutical dosage form is characterized
by being
bioequivalent when administered orally, fasted, in a hard capsule shell
compared to the beads
-14-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
being administered orally, fasted, without a capsule shell. For example, the
pharmaceutical
dosage form can be characterized by the dosage form when administered orally
in a hard
capsule shell exhibiting a Cmax in a range of 80% to 125%, or 80% to 120%, of
Cmax
exhibited by the beads administered orally without a capsule shell. In another
embodiment,
the dosage form can be characterized by the dosage form when administered
orally in a hard
capsule shell exhibiting an AUC (0-12h) or AUC (0-inf) in a range of 80% to
125%, or 80%
to 120%, of that exhibited by the beads administered orally without a capsule
shell,
respectively. In one embodiment, both the Cmax and the AUC are within the
tolerance
ranges just described.
[0068] Purity
[0069] In one type of embodiment, the dosage form is characterized by having
less than 5
wt.% cystamine, based on the amount of cysteamine, as determined by reverse
phase HPLC
with UV detection, as described herein. In other embodiments, the dosage form
is
characterized by having less than 5 wt.% cystamine, based on the amount of
cysteamine,
following 12 months storage at 25 C and 40% relative humidity (RH),
optionally as
determined by reverse phase HPLC with UV detection, as described herein. In
another type
of embodiment, the dosage form is characterized by having less than 5 wt.%
cystamine,
based on the amount of cysteamine, following 18 months storage at 25 C and
40% RH
optionally as determined by reverse phase HPLC with UV detection, as described
herein. In
another type of embodiment, the dosage form is characterized by having less
than 5 wt.%
cystamine, based on the amount of cysteamine, following 24 months storage at
25 C and
40% RH optionally as determined by reverse phase HPLC with UV detection, as
described
herein. In another type of embodiment, the dosage form is characterized by
having less than
wt.% cystamine, based on the amount of cysteamine, following 30 months
storage, or more,
at 25 C and 40% RH optionally as determined by reverse phase HPLC with UV
detection, as
described herein. Examples of suitable reverse phase HPLC assays are described
herein.
[0070] In another type of embodiment, the dosage form is characterized by
having less
than 5 wt.% cystamine, based on the amount of cysteamine, following 12 months
storage at
25 C and 60% RH, optionally as determined by reverse phase HPLC with UV
detection, as
described herein. In another type of embodiment, the dosage form is
characterized by having
less than 5 wt.% cystamine, based on the amount of cysteamine, following 18
months storage
at 25 C and 60% RH, optionally as determined by reverse phase HPLC with UV
detection,
as described herein. In another type of embodiment, the dosage form is
characterized by
having less than 5 wt.% cystamine, based on the amount of cysteamine,
following 24 months
-15-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
storage, or more, at 25 C and 60% RH, optionally as determined by reverse
phase HPLC
with UV detection, as described herein.
[0071] In another type of embodiment, the dosage form is characterized by
having less
than 5 wt.% cystamine, based on the amount of cysteamine, following 3 months
storage at 40
C and 75% RH, optionally as determined by reverse phase HPLC with UV
detection, as
described herein. In another type of embodiment, the dosage form is
characterized by having
less than 5 wt.% cystamine, based on the amount of cysteamine, following 6
months storage
at 40 C and 75% RH, optionally as determined by reverse phase HPLC with UV
detection,
as described herein.
[0072] Any of the foregoing embodiments can be further characterized by having
less than
8 wt.% total related substances (impurities) based on the amount of
cysteamine, under the
described storage conditions and times based on reverse phase HPLC with UV
detection, as
described herein.
[0073] Method of making
[0074] Also contemplated is a method for the preparation of a dosage form
according to
the disclosure here, including coating a core particle comprising cysteamine
or a
pharmaceutically acceptable salt thereof and an excipient with an enteric
polymer to form the
enteric membrane.
[0075] The core particle including cysteamine or a pharmaceutically acceptable
salt thereof
can be formed by any suitable process. In one embodiment, the core particle is
formed by
granulating a mixture of cysteamine or a pharmaceutically acceptable salt
thereof with an
excipient and milling to a desired particle size range. In another embodiment,
the core
particle can be formed by extrusion and spheronization of a mixture of
cysteamine or a
pharmaceutically acceptable salt thereof with an excipient. Granulating
processes can
include fluid bed granulation, wet granulation, hot melt granulation, and
spray congealing, for
example. Other processes include slugging and roller compaction. As it is
known in the art,
the mixtures which are to be granulated can first be dry-blended. The dry-
blended dry
ingredients can be mixed with water, prior to extrusion.
[0076] It has been found that extrusion and spheronization of a mixture of
cysteamine or a
pharmaceutically acceptable salt thereof with an excipient can provide
desirable core particles
with a distribution of particle sizes as described herein and one or more
other desirable
properties. Cysteamine bitartrate oxidizes in air and in water, and with heat.
Thus, short
processing times can lead to a more stable product. For example, reducing the
amount of
-16-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
spheronization reduces the amount of friction and related heat. For example,
reducing the
amount of time that the product is exposed to air (either in the moist state
and/or before
packaging) also reduces the amount of oxidation. On the other hand, rapid
processing by
extrusion and spheronization can lead to a poor quality product, for example
in having a large
fraction of the pellet cores falling outside a desired particle size range.
The amount of
moisture absorbed by spheronization aids (which does not happen immediately,
but instead
over time) influences the spheronization characteristics of the beads.
Accordingly, it was
determined that the moisture content of the wet mass, the related wet hold
time for swelling
of spheronization aid(s), and the spheronization time are parameters that can
be optimized to
achieve both good product yield, for example in a particle size range
described herein, while
maintaining good stability, e.g. not more than 5 wt.% cystamine based on the
amount of
cysteamine, as described herein.
[0077] Accordingly, in one embodiment the moisture content of the granulation
mixture,
prior to drying, is in a range of about 20 wt.% to about 40 wt.%, or 25 wt.%
to about 35
wt.%, or about 28 wt.% to about 32 wt.%, or at least about 28 wt.%, or at
least about 28.5, or
at least about 20 wt.% to about 40 wt.%, or at least about 25 wt.% to about 35
wt.%, or at
least about 27 wt.% to about 31 wt.% or at least about 28.5 wt.% to about 31
wt.%.
[0078] The wet mass can be held for a period of time prior to extrusion, e.g.
in order to
allow the sphronization aid to swell with granulating fluid. The hold time can
be at least 15
minutes, at least 30 minutes, at least 45 minutes, or at least 60 minutes, for
example. The
hold time can be in a range of about 15 minutes to about 120 minutes, or about
30 minutes to
100 minutes, or 60 minutes to 90 minutes, for example.
[0079] As described above in connection with description of the core
particles, the method
can include a step of sorting (e.g., by sieving) the core particles prior to
enteric coating, to
retain particles in a predetermined size range, for example sizes in a range
of about 0.7 mm to
about 2.8 mm, or about 0.7 mm to about 2.5 mm, or about 0.8 mm to about 1.7
mm, or any
range described above in connection with the core particles.
[0080] As described above in connection with description of the beads, the
method can
include a step of sorting (e.g., by sieving) the beads after enteric coating,
to retain particles in
a predetermined size range, for example sizes in a range of about 0.7 mm to
about 2.8 mm, or
about 0.7 mm to about 2.5 mm, or about 0.8 mm to about 1.7 mm, or any range
described
above in connection with the core particles.
-17-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0081] In an extrusion and spheronization process, the following optional
features can be
employed, individually or in one or more combinations thereof. Water can be
used as a
granulation agent. Microcrystalline cellulose can be used in the core
particles as a
spheronization aid. Hypromellose can be included in the core particles as a
binder. The
extrusion screen size can be 1.0mm. The friction plate of the spheronizer can
be cross-
hatched. The friction plate of the spheronizer can be cross-hatched with a
square pitch of at
least 3mm, or greater than 3 mm, or at least 4mm, or greater than 4mm, or in a
range of about
3 mm to about 7 mm, or about 5 mm. The spheronization time can be less than
about 5
minutes, or less than about 4 minutes, or less than about 3 minutes, or less
than about 2
minutes, or up to 1 minute. The spheronized particles can include non-
spherical particles (i.e.
irregular shapes), e.g. a substantial fraction thereof, e.g. at least 20 wt.%
or at least 30 wt.%,
or at least 40 wt.% or at least 50 wt.% or at least 60 wt.%, or at least 70
wt.% thereof.
[0082] The beads and/or filled capsules can be stored with a desiccant. The
beads and/or
filled capsules can be stored with an oxygen absorber.
[0083] For example, one embodiment of the method combining various of the
parameters
described above includes a method for the preparation of a pharmaceutical
dosage form
including cysteamine beads, including forming a wet mass comprising cysteamine
bitartrate
and an excipient, optionally microcrystalline cellulose, with a moisture
content in a range of
in a range of about 20 wt.% to about 40 wt.%, extruding and spheronizing the
wet mass
including cysteamine bitartrate and excipient to make core particles, sorting
the core particles
to a target particle size range, optionally 0.7 mm to 2.5 mm, coating the
sorted core particles
with an enteric polymer to form including beads comprising a core particle and
an enteric
membrane, and sorting the bead particles to a target particle size range,
optionally 0.7 mm to
2.5 mm.
[0084] Use / Administration
[0085] For administration of the dosage form, a total weight in the range of
approximately
100 mg to 1000 mg (based on the free base) can be used. The dosage form can be
orally
administered to a patient suffering from a condition for which an cysteamine
is indicated,
including, but not limited to, cystinosis and other metabolic and
neurodegenerative diseases
including non-alcoholic fatty liver disease (NAFLD), Huntingon's disease,
Parkinson's
disease, Rett Syndrome and others, use as free radical and radioprotectants,
and as hepto-
protectant agents. In any method described herein, the treatment of humans is
contemplated.
The compositions of the disclosure can be used in combination with other
therapies useful for
-18-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
treating cystinosis and neurodegenerative diseases and disorders. For example,
indomethacin
therapy (Indocid or Endo110) is an anti-inflammatory used to treat rheumatoid
arthritis and
lumbago, but it can be used to reduce water and electrolyte urine loss. In
children with
cystinosis, indomethacin reduces the urine volume and therefore liquid
consumption by about
30%, sometimes by half. In most cases this is associated with an appetite
improvement.
Indomethacin treatment is generally followed for several years.
[0086] Other therapies can be combined with the methods and compositions of
the
disclosure to treat diseases and disorders that are attributed or result from
cystinosis. Urinary
phosphorus loss, for example, entails rickets, and it may be necessary to give
a phosphorus
supplement. Carnitine is lost in the urine and blood levels are low. Carnitine
allows fat to be
used by the muscles to provide energy. Hormone supplementation is sometimes
necessary.
Sometimes the thyroid gland will not produce enough thyroid hormones. This is
given as
thyroxin (drops or tablets). Insulin treatment is sometimes necessary if
diabetes appears,
when the pancreas does not produce enough insulin. These treatments have
become rarely
necessary in children whom are treated with cysteamine, since the treatment
protects the
thyroid and the pancreas. Some adolescent boys require a testosterone
treatment if puberty is
late. Growth hormone therapy may be indicated if growth is not sufficient
despite a good
hydro electrolytes balance. Accordingly, such therapies can be combined with
the
compositions and methods disclosed herein.
[0087] The effectiveness of a method or composition of the disclosure can be
assessed by
measuring leukocyte cystine concentrations. Dosage adjustment and therapy can
be made by
a medical specialist depending upon, for example, the concentration of cystine
in leukocytes
and the ability to tolerate the drug. Additional therapies including the use
of omeprazole
(Priloseci0) can reduce side effects of cysteamine administration, such as
abdominal pain,
heartburn, nausea, vomiting, and anorexia, which can result from cysteamine-
induced gastric
acid hypersecretion, for example.
[0088] In addition, various prodrugs can be "activated" by use of the
enterically coated
cysteamine. Prodrugs are pharmacologically inert, they themselves do not work
in the body,
but once they have been absorbed, the prodrug decomposes. The prodrug approach
has been
used successfully in a number of therapeutic areas including antibiotics,
antihistamines and
ulcer treatments. The advantage of using prodrugs is that the active agent is
chemically
camouflaged and no active agent is released until the drug has passed out of
the gut and into
the cells of the body. For example, a number of prodrugs use S¨S bonds. Weak
reducing
agents, such as cysteamine, reduce these bonds and release the drug.
Accordingly, the
-19-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
compositions of the disclosure are useful in combination with pro-drugs for
timed release of
the drug. In this aspect, a pro-drug can be administered followed by
administration of an
enterically coated cysteamine composition of the invention (at a desired time)
to activate the
pro-drug.
EXAMPLES
[0089] The following examples are provided for illustration and are not
intended to limit
the scope of the invention.
Example 1 ¨ Bead Production
[0090] Cysteamine bitartrate and excipients (microcrystalline cellulose,
hypromellose,
sodium lauryl sulfate) were milled through a Comil equipped with a 0.094"
(2.3876 mm)
screen operating at 500 RPM. The amount of each ingredient (per 75mg
cysteamine capsule)
is cysteamine bitartrate 258mg +/- 37.0 mg; microcrystalline cellulose 67.1mg
+/- 9.6 mg;
hypromellose 17.2mg +/- 2.5 mg; and sodium lauryl sulfate 1.75 mg +/ 0.25 mg.
Cysteamine
bitartrate was passed through the Comil first followed by the excipients
(hypromellose 2910-
5, sodium lauryl sulfate, and microcrystalline cellulose). Cysteamine
bitartrate and the
excipients were dry blended for approximately 15 minutes. While mixing at a
setpoint speed
of 47 rpm, purified water was slowly added (addition in approximately 4
minutes) into the
blended components. After the water addition, the wet blend was mixed for an
additional
minute for a total of 5 minutes.
[0091] A sample of the wet blend was collected and moisture content was
determined by
loss on drying (LOD). The wet mass was discharged in polyethylene lined fiber
drums and
held for 60-90 minutes prior to extrusion/spheronization.
[0092] The granulated wet mass was loaded onto a NICA extruder equipped with a
1.0 mm
screen at a feeder speed of 100 RPM setpoint and extruded at a setpoint speed
of 55 RPM
(50-60 RPM). The extruded product was immediately spheronized using a NICA
Spheronizer equipped with 5.0 mm cross-hatched friction plates. Spheronization
was
performed at a target speed of 625 RPM (500-700 RPM) for 40-60 seconds. The
particles
were collected in double polyethylene lined fiber drums and stored at room
temperature for
further processing.
[0093] The wet particles were dried in a Niro fluid bed dryer with an inlet
air temperature
setpoint of 70 C (60-80 C). Drying was complete when the moisture content of
uncrushed
particles reached <1.0% w/w by LOD. Sampling of the particles began when the
outlet air
temperature reached approximately 50 C and continued until the acceptance
criterion of
-20-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
<1.0%. The dried particles were transferred to fiber drums lined with double
polyethylene
bags and stored at room temperature.
[0094] The dried particles were screened through a #12 mesh screen and a #20
mesh
screen. Particles passing through the #12 mesh and retained on the #20 mesh
were collected
as product in double polyethylene lined containers with desiccant and oxygen
absorber
packets in the outer liner. The collected product may be re-passed through the
screens as
needed. Particles greater than #12 mesh and less than #20 mesh were not
retained as product
for coating.
[0095] An enteric coating solution of Eudragit L30 D-55, triethyl citrate, and
talc in
purified water was prepared in a mixing tank equipped with a propeller mixer
and placed on a
balance. Eudragit L 30 D-55 was added to the portable mixing tank through a 60-
mesh
screen. The final solution was mixed for a minimum of 30 minutes and mixed
continuously
during the coating process. Based on a 75mg cysteamine capsule, the amounts of
coating
ingredients were: Eudragit L30 D-55 66.2 mg +/- 9.5 mg; triethyl citrate 6.65
mg +/- 0.95
mg; talc 15.3 mg +/- 2.2 mg.
[0096] Spray lines connecting the portable mixing tank to the Niro fluid bed
dryer were
primed. The floor balance was tared prior to starting the coating process. The
amount of
coating solution sprayed was calculated as the amount required to increase the
core particle
weight by 25%.
[0097] The core particles were loaded into the Niro fluid bed dryer equipped
with a
Precision Coater which sprays from the bottom, 1.0 mm Nozzle, 30 mm Swirl
Accelerator,
and 300[1.m Filter Bonnet. The coating process parameters are provided in the
table below.
Parameter Setpoint Range
Inlet Air Volume 450 scfm 300-600 scfm
Inlet Air Temperature 60 C 45 - 75 C
Product Temperature 30 C 25 - 45 C
Solution Spray Rate 0.220 kg/minute 0.200 ¨ 0.240 kg/minute
Atomization Air Pressure 36 psi 32-40 psi
[0098] Once the target weight of coating solution was applied (25% of dry
particle
weight), the beads were weighed to confirm weight increase of >25.0%. If the
weight was
not >25.0% of the uncoated particle weight, the coating process was continued
until >25.0%
was achieved.
-21-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0099] The coated beads were dried at an inlet temperature setpoint of 45 C
(35 - 55 C)
and inlet air volume setpoint of 350 scfm (300-400 scfm) until the LOD of the
coated beads
was <2.0% w/w. Once the LOD was reached, the inlet air heating was turned off
and the
beads were circulated at an air inlet volume of 300-400 scfm until the product
temperature
reached not more than (NMT) 30 C.
[0100] The weight gain of the dried coated beads was calculated to confirm a
maximum
weight gain of <31.0% was achieved. Visual inspection confirmed that the
enteric membrane
thickness was not consistent bead-to-bead, but instead there was a
distribution of enteric
membrane thicknesses.
[0101] The dried coated beads were screened through a #12 mesh and a #20 mesh
screen
in sequence. Beads passing through the #12 mesh screen and retained on the #20
mesh
screen were collected as product in double polyethylene lined fiber drums with
a desiccant
and oxygen absorber canister in the outer liner. Mesh analysis testing can be
performed as an
in-process test to confirm the beads are within the limits of: NMT 5% are
retained on a #12
mesh screen (1.68 mm) and NMT 10% pass through a #20 mesh screen (0.84 mm). If
results
are not within the limits, the product can be sorted by rescreening until the
mesh analysis
results meet the specified limits.
[0102] The dried coated beads were lubricated with talc prior to
encapsulation. The coated
beads were loaded in a V- blender; talc powder was added to the coated beads
(calculated as
0.5% w/w of the total coated bead weight). The contents were mixed for a
minimum of five
minutes. The lubricated coated beads were transferred to double polyethylene
lined fiber
drums with desiccant and oxygen absorber packets in the outer liner and stored
at room
temperature. Lubricated coated beads were used in the manufacture of 75 mg
size 0 capsules
and 25 mg size 3 capsules. One batch of coated beads can be filled as a 75 mg
strength batch
or can be split to fill both 75 mg and 25 mg strengths, for example.
[0103] The 75 mg hard gelatin capsules were filled using an automated
encapsulator at a
speed of 80 - 100 spm to the target fill weight calculated to achieve 75 mg
cysteamine free
base per capsule. The 25 mg hard gelatin capsules were also filled with an
automated
encapsulator at a speed of 50-70 spm. The beads were introduced into the
encapsulation
process with a hopper.
Example 2 ¨ Particle Size Distribution
-22-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0104] Several lots of cysteamine bitartrate enteric-coated beads produced via
an extrusion
and spheronization process as described herein were analyzed for particle size
distribution via
analytical sieving. The results are tabulated below.
% Retained % Retained % Retained % Retained
Sieve Size (1J m) Lot A Lot B Lot C Lot D
1700 0 0 0 0
1400 1.4 3.2 3.2 1.2
1180 19.5 25.7 26.7 20.3
1000 61.9 55.5 56 62
850 16.1 14.2 13.5 15.1
<850 1.2 1.4 0.6 1.4
Example 3 - Pharmacokinetics
[0105] A population PK study was performed using Cystagon and capsules of
cysteamine
bitartrate gastro-resistant beads (CBGB) produced according to the method of
Example 1
herein.
[0106] Pharmacokinetic (PK) and pharmacodynamic (PD) relationships following a
single
dose of CBGB capsules was first studied in comparison to a single dose of
immediate-
release cysteamine bitartrate in a study with 9 patients. Following
normalization to a 450 mg
dose, the maximum plasma levels C max , AUC 0-6h and AUC 0-12h (calculated
directly
from the plasma level data for CBGB and from doubling the AUC 0-6h value for
immediate-
release cysteamine to represent two doses) were lower for CBGB (27.70 14.99
[tmol/L,
75.93 39.22 p.mol*h/L and 99.26 44.21 p.mol*h/L respectively) than for
immediate-
release cysteamine bitartrate (37.72 12.10 [tmol/L, 96.00 37.81 p.mol*h/L
and 192.00
75.62 p.mol*h/L respectively. The pharmacokinetics of CBGB are consistent with
a delayed-
release formulation showing a T max of 2.78 1.56 h for CBGB cysteamine was
moderately bound to human plasma proteins, predominantly to albumin, with mean
protein
binding of about 52%. Plasma protein binding was independent of concentration
over the
concentration range achieved clinically with the recommended doses.
[0107] Additional studies were carried out as follows.
[0108] CBGB-A Study
[0109] Cystagon Treatment Assignment: one (1) pre-dose PD sample was
collected at
time 0 (i.e., within 15 minutes prior to the morning Cystagon dose
administration),
considered as the time of trough cysteamine / peak of WBC cystine after
administration of
-23-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
immediate-release cysteamine bitartrate (Cystagon ). One (1) additional PD
sample was
collected at a sample timepoint that was time-matched to 1 of 3 PK sample
profile times
(either 2, 4 or 6 hours) post morning Cystagon dose. There were six
associated plasma PK
samples collected at time 0 (within 15 minutes prior to morning Cystagon
dose); 30 minutes
post morning Cystagon dose; and 1, 2, 4 and 6 hours (immediately prior to the
afternoon
Cystagon dose)
[0110] Inventive capsule Treatment Assignment: one (1) post-dose PD sample was

collected at time 0.5 hour (30 minutes), considered as the time of trough
cysteamine / peak of
WBC cystine after administration of capsules of CBGB. Two (2) additional PD
samples
were collected at sample timepoints that were time-matched to PK sample
profile times
(either 3, 4, 8, 10 or 12 hours) post morning CBGB dose. In order to limit the
impact of
autocorrelation, juxtaposed times of sampling for patients treated with CBGB
were not to be
taken into account for the randomization. Therefore, patients were randomized
to one of the
following six pairs of the sampling time points: 3 and 8 hours, 3 and 10
hours, 3 and 12
hours, 4 and 8 hours, 4 and 10 hours, 4 and 12 hours. There were nine
associated plasma PK
samples collected at time 0 (within 15 minutes prior to morning CBGB dose), 30
minutes, 2,
3, 4, 6, 8, 10 and 12 hours post morning CBGB dose (immediately prior to the
evening
CBGB dose).
[0111] As recommended in the Cystagon SmPC, food (meal or snack) was
available 30
minutes prior to receiving the morning dose and (if applicable) the next Q6H
of Cystagon
administration and the morning dose and Q 12H CBGB administration and (if
applicable) the
next Q12H CBGB dose. Cystagon was administered with water and CBGB was
administered with an acidic beverage. Dairy products should have been withheld
1 hour
before and after CBGB dosing.
[0112] CBGB-B Study
[0113] Administering cysteamine in fasted healthy volunteers provides very
stable PK
parameters such that it was possible to demonstrate bioequivalence between
administrations
of CBGB capsules as a whole or as their content sprinkled on food with only 20
healthy
volunteers.
[0114] The PK parameters of cysteamine were determined after a single dose,
first in
fasted healthy volunteers, then in patients at steady state, using the model
parameters
obtained with healthy volunteers as starting parameters for the models in
patients.
Pharmacokinetic modeling of cysteamine was based on a 2-compartment model and
-24-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
pharmacodynamic modeling of WBC cystine was based on an inhibitory Emax model.

(Belldina, E. B., M. Y. Huang, et al. (2003). "Steady-state pharmacokinetics
and
pharmacodynamics of cysteamine bitartrate in paediatric nephropathic
cystinosis patients." Br
J Clin Pharmacol 56(5): 520-525.)
[0115] Since CBGB studies in healthy volunteers were not done against Cystagon
, data in
fasted healthy volunteers (Gangoiti, J. A., M. Fidler, et al. (2010).
"Pharmacokinetics of
enteric-coated cysteamine bitartrate in healthy adults: a pilot study." Br J
Clin Pharmacol
70(3): 376-382) were used to determine initial PK model parameters for
Cystagon . And data
on EC-cysteamine (i.e. Eudragit L5OD 55 enteric-coated capsules of Cystagon -
a different
way of providing delayed-release cysteamine bitartrate) in this dataset was
used for
comparison purposes.
[0116] A bioequivalence designed to demonstrate bioequivalence between oral
administration of intact CBGB capsules, and contents of opened CBGB capsules
mixed with
applesauce and taken orally. Twenty (20) healthy adults (mean age 37 years,
range 19-64
years) received both presentations, 8 (75 mg) intact vs. 8 (75 mg) open
capsules, in a
crossover design study.
[0117] The final results are presented in the table below.
-25-

0
n.)
o
1-,
.6.
Study/ Study No. Subjects HV/Pa) Treatment Dose Non-
Compartmental Analysis Population PK, 2-compartment Model
.6.
Protocol Design Entered/ (Age: Mean, (mg) (Pharsight,
WinNonLin 6.2) (Pharsight, NLME 1.1) oec'e
Country Completed Range)
(M/F)
Tmax Cmax Cmax D
AUCinf D Tiag Ka V/F Cl/F V2 C12
(min) (mg/L)
(mg/L/mg) (min*mg/L/mg) (min) (1/min) (L) (L/min) (L) (L/min)
UCSD Open label, (4M/3F)/ P (12, 8-17) Cystagon 450
75 3.1 0.007 0.88 0.30 26 0.029 73 1.07
131 0.41
(USA) Sequential (4M/3F) 19 1.2 0.003
EC- 450 220 3.2
0.007 0.96 0.40 156 0.025 98 1.17 54 0.5
Cysteamine 74 1.4 0.003
P
t.)
.
'? CBGB-A Random, (24M/19F)/ P(12,6-26) Cystagon 250-
750 74 32 2.6 0.006 0.84 0.31 23 0.025 94 1.1
191 0.5
(USA/
1-
Crossover (22M/16F) 1.4 0.003
0.
..]
_.,
EU) CBGB caps 425- 183 3.5
0.005 1.08 0.46 60 0.015 87 1.2 200 0.4
0
1-
1300 90 1.7 0.002
u,
1
1-
IV
,
CBGB-B Random, (13M/7F)/ HV(37, 19- CBGB caps 600 194
2.3 0.004 0.84 0.19 95 0.016 137 1.4 187
0.44 0
..J
(USA) Crossover (13M/7F) 64) 38 0.6 0.001
CBGB 600 190 2.3
0.004 0.85 0.21 98 0.017 151 1.4 192 0.47
sprinkled 61 0.7 0.001
a) HV = Healthy Volunteers, P = Patients
IV
n
cp
t..,
=
.6.
-a-,
.6.
t..,
cA
=
--.1

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0118] The conclusion of this population PK modeling on two different
presentations of
CBGB (open and intact), is that the only difference between administering CBGB
as intact
capsules and as open capsules, sprinkled on applesauce, is expressed by the
difference
between lag times: as expected the start of absorption from the beads is still
delayed (85 min)
but slightly less than when the gelatin capsule has to be dissolved first (108
min) and this has
not much of an impact on Tma, (190 min for open capsules vs. 194 min for
intact capsules)
since probably only a small amount of beads dissolves early.
[0119] However, comparison between the two presentations of CBGB (open and
intact)
and the immediate-release cysteamine bitartrate (Cystagon ) and the delayed-
release EC-
cysteamine, shows that the absorption of cysteamine after CBGB dosing is not
only more
delayed (Cystagon Tiag << CBGB Tiag << EC-cysteamine Tiag) but also further
extended due
to a slower absorption (CBGB Ka << Cystagon Ka z EC-cysteamine Ka) compared
to EC-
cysteamine. Without intending to be bound by any particular theory, it is
contemplated that
the difference in absorption of the CBGB formulation is related to one or more
factors
including the distribution of bead sizes and time-progressive dissolution of
multiple beads
and/or the irregularity of bead shapes in the CBGB formulation and/or the
distribution of
enteric membrane thicknesses in the CBGB formulation.
Example 4 ¨ Purity and Stability
[0120] Long term stability tests have been performed on the CBGB formulation
made
according to Example 1. The major impurity in the CBGB product is cystamine,
the well
known related substance (dimer).
[0121] The use of a more sensitive and less selective method has resulted in
the
observation of several impurities found in the CBGB formulation and the
commercial product
using cysteamine bitartrate, Cystagon . Through the use of reverse phase HPLC,
six peaks
observed in the CBGB formulation related substances chromatograms have been
identified as
product degradants (specifically cysteamine bitartrate degradants). Two lots
of Cystagon
were evaluated by the same test method. The impurities observed in
representative CBGB
chromatograms are also observed in Cystagon .
[0122] Impurities Assay Method
[0123] Cysteamine bitartrate samples are assessed by gradient elution HPLC
using an
XBRIDGE C18 column (dimensions: 150 mm x 4.6 mm; packing particle size: 3.5
p.m)
(Waters, Milford, Massachusetts). The autosampler temperature is 4 C.
Approximately 10
[IL or approximately 100 [IL of sample is injected onto the column. The column
temperature
-27-

CA 02914770 2015-12-07
WO 2014/204881
PCT/US2014/042607
is 40 C and the sample is eluted at a flow rate of 1.0 mL/min according to the
following
profile:
HPLC Gradient
Time (min) Mobile Phase A (%) Mobile Phase B (%)
0.0 100 0
2.0 100 0
20.0 60 40
25.0 60 40
25.1 100 0
40.0 100 0
[0124] Mobile Phase A contains 23.6 mM 1-octanesulfonic acid sodium and 29.0
mM
sodium phosphate (pH 2.6)/acetonitrile/methanol 85/3/12 (v/v/v). Mobile Phase
B contains
0.20 M 1-octanesulfonic acid sodium and 0.10 M sodium phosphate (pH
2.6)/acetonitrile/methanol 10/18/72 (v/v/v). The purity of 1-octanesulfonic
acid is > 98%.
Detection is carried out using a UV detector at 210 nm.
[0125] Reference Solution Preparation. Reference solutions of Cysteamine
Bitartrate
Analytical Reference Standard are prepared as follows. Working Standard and
Working
Check Standard solutions are prepared having a nominal concentration of 0.54
mg/mL
Cysteamine Bitartrate Analytical Reference Standard in Mobile Phase A using
low actinic
glassware. A Working Sensitivity solution is prepared having a nominal
concentration of
0.30 mg/mL Cysteamine Bitartrate Analytical Reference Standard in Mobile Phase
A using
low actinic glassware, which corresponds to the limit of quantification (LOQ)
for cysteamine.
The water content of the Cysteamine Bitartrate Analytical Reference Standard
is determined
no more than 7 days before use by Karl Fischer titration or thermal
gravimetric analysis
(TGA). The Reference Standard is stored refrigerated and blanketed under
nitrogen.
[0126] Bead Prep Assay Sample Preparation. Cysteamine Bitartrate Gastro-
resistant
Beads (CBGB) are prepared for analysis according to the following procedure.
About 3.7 g
of CBGB beads are ground to a fine powder using a ball mill for approximately
1 minute at
27 Hz. The grind is transferred to an amber bottle for storage. Stock Bead
Prep Assay
sample solutions are prepared in duplicate by adding 370.4 mg 5 mg of the
grind to a 250
mL low actinic volumetric flask and diluting with Mobile Phase A. The mixture
is stirred
with a stir bar for at least 15 minutes. Approximately 15 mL of the resulting
solution is
filtered through a 0.45 lam nylon filter, with the first 5 mL being discarded.
The cysteamine
concentration of the resulting Stock Bead Prep Assay sample solution is
approximately 0.300
-28-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
mg/mL. Working Bead Prep sample solutions are prepared by placing 4.0 mL of
Stock Bead
Prep Assay sample solution in a 25 mL low actinic volumetric flask and
diluting to volume
with Mobile Phase A. The cysteamine concentration of the resulting Working
Bead Prep
sample solution is approximately 0.048 mg/mL.
[0127] Assay Sample Preparation. CBGB capsules are prepared for analysis
according
to the following procedure. To reduce exposure to light and oxygen, sample
preparation
(from the initial weighing of the full capsules to the loading of sample vials
on the HPLC) is
completed in one day. Ten capsules are weighed. The capsule contents are
emptied and the
empty shells are weighed to determine the average capsule fill weight. The
capsule contents
are ground to a fine powder using a ball mill for approximately 1 minute at 27
Hz. The grind
is transferred to an amber bottle for storage. Stock sample solutions are
prepared in duplicate
by adding the appropriate amount of the grind for 1 capsule (as determined by
the average
capsule fill weight) to a 25 mL low actinic volumetric flask and diluting with
Mobile Phase
A. The mixture is stirred with a stir bar for at least 15 minutes. The
resulting solution is
centrifuged at about 3400 rpm for 5 minutes. Approximately 15 mL of the
centrifuged
solution is filtered through a 0.45 [tm nylon filter (Acrodisc, 25 mm
diameter), with the first 5
mL being discarded, to obtain Stock sample solutions. Working sample solutions
are
prepared by placing 6.0 mL of Stock sample solution (for 25 mg capsules) or
2.0 mL of Stock
sample solution (for 75 mg capsules) in a 10 mL low actinic volumetric flask
and diluting to
volume with Mobile Phase A.
[0128] Content Uniformity Sample Preparation. CBGB capsules are prepared for
analysis according to the following procedure. To reduce exposure to light and
oxygen,
sample preparation (from the initial weighing of the full capsules to the
loading of sample
vials on the HPLC) is completed in one day. Ten capsules are weighed. The
contents of each
capsule are emptied into separate mortars and the empty shells are weighed to
determine the
individual capsule fill weight. About 1-2 mL of Mobile Phase A is added into
the mortar.
The beads are immediately ground to a paste. If needed, additional Mobile
Phase A is added
to the paste, up to 5 mL total. The paste is transferred to a 250 mL low
actinic volumetric
flask. The mortar and pestle are thoroughly rinsed with Mobile Phase A and the
rinse
solution is collected in to the same flask. The flask is filled about three-
quarters full with
Mobile Phase A and stirred for at least 15 minutes. The flask is filled to
volume with Mobile
Phase A. Approximately 20 mL of the resulting solution is filtered through a
0.45 [tm nylon
filter (Acrodisc, 25 mm diameter), with the first 5 mL being discarded, to
obtain Stock CU
sample solutions. Working CU sample solutions are prepared by placing 12.0 mL
of Stock
-29-

CA 02914770 2015-12-07
WO 2014/204881
PCT/US2014/042607
CU sample solution (for 25 mg capsules) or 4.0 mL of Stock CU sample solution
(for 75 mg
capsules) in a 25 mL low actinic volumetric flask and diluting to volume with
Mobile Phase
A. The cysteamine concentration of the resulting Working CU sample solutions
is
approximately 0.048 mg/mL.
[0129] Data Analysis. The cysteamine Working Standard solution concentration
is
calculated according to the following equation: Cysteamine Concentration
(Cstd) =
mg Cysteamine Bitartrate Analytical Reference Standard x Pf / 25.0 mL
[0130] Pf represents a purity factor for the standard material. Pf is
calculated according to
the following equation: Pf = B x (100-Water) x C / 100
where B = the anhydrous cysteamine free base in the Cysteamine Bitartrate
Analytical
Reference Standard (expressed as a decimal value on the standard bottle
label),
water = the water content as determined by Karl Fischer or TGA no more than 7
days before
use (expressed as a percentage), and
C = the cystamine correction (expressed as a decimal value on the standard
bottle label).
[0131] The amount of cysteamine per capsule is calculated according to the
following
equation: mg cysteamine per capsule =
(Asam/Astd) X Cstd X DF x (AveWt/SamWt)
where Asam = the peak area of cysteamine in the sample chromatogram with a 10
[t.L
injection,
Astd = the average peak area of cysteamine in all Working Standard solution
chromatograms
with a 10 [t.L injection,
CStd = the concentration (mg/mL) of cysteamine in the Working Standard
solution,
DF = the dilution factor (125 for 75 mg capsules; 41.6667 for 25 mg capsules),
AveWt = the average capsule fill weight (mg), and
SamWt = the sample weight (mg).
[0132] For Content Uniformity, the amount of cysteamine per capsule is
calculated
according to the following equation: mg cysteamine per capsule =
(Asam/Astd) X CStd X DF
where Asam = the peak area of cysteamine in the sample chromatogram with a 10
[t.L
injection,
Astd = the average peak area of cysteamine in all Working Standard solution
chromatograms
with a 10 [t.L injection,
-30-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
CStd = the concentration (mg/mL) of cysteamine in the Working Standard
solution, and
DF = the dilution factor (1562.5 for 75 mg capsules; 520.8 for 25 mg
capsules).
[0133] For the Bead Prep Assay, the amount of cysteamine per capsule is
calculated
according to the following equation: mg cysteamine per capsule =
(Asam/Astd) X Cstd X DF x (AveWt/SamWt)
where As. = the peak area of cysteamine in the sample chromatogram with a 10
[t.L
injection,
AStd = the average peak area of cysteamine in all Working Standard solution
chromatograms
with a 10 [t.L injection,
Cstd = the concentration (mg/mL) of cysteamine in the Working Standard
solution,
DF = the dilution factor (use the 75 mg Dilution Factor, 1562.5),
AveWt = the average capsule fill weight (mg) (use the target fill weight,
370.4 mg), and
SamWt = the sample weight (mg) (use the actual weight used in sample
preparation).
[0134] The percentage of the label claim (%LC) is calculated for the Assay,
Content
Uniformity, and Bead Prep Assay sample solutions according to the following
equation:
%LC = (mg cysteamine)/LC x 100%
where mg cysteamine = the amount calculated by the applicable equation above,
and
LC = the amount of the label claim (75 mg or 25 mg) (use 75 mg for the Bead
Prep Assay).
[0135] The amount of substances related to cysteamine bitartrate (including
cysteamine
impurities) such as cystamine is calculated according to the following
equation:
mg related substance = (ARs/Astd) x (Cstd/RRF) x DF x (AveWt/SamWt)
where ARs = the peak area of any related substance in the Working sample
solution
chromatogram with a 100 [t.L injection (peaks before RRT 0.48 are disregarded;
peaks
observed in the chromatogram of the second injection of Mobile Phase A/Blank
(100 [t.L
injection) are also disregarded),
AStd = the average peak area of cysteamine in all Working Standard solution
chromatograms
with a 10 [t.L injection,
Cstd = the concentration (mg/mL) of cysteamine in the Working Standard
solution,
RRF = the relative response factor (0.98 for cystamine; 1.00 for other related
substances),
DF = the dilution factor (12.5 for 75 mg capsules; 4.16667 for 25 mg
capsules),
AveWt = the average capsule fill weight (mg), and
SamWt = the weight the sample grind from the Working sample solution
preparation (mg).
-31-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
[0136] The weight percentage of cystamine and other individual related
substances is
determined according to the following equation:
% individual related substance = mg related substance / mg cysteamine x 100%
where mg related substance = the amount of related substance calculated above,
and
mg cysteamine = the amount of cysteamine for the Assay sample.
[0137] The percentage of total related substances is determined by summing all
related
substances greater than or equal to 0.05%. Peaks after 28 minutes are
disregarded. In
contrast to a previous electrochemical detection method that disregarded early-
eluting peaks
as not relevant to the purity calculation, the foregoing method determines
that early peaks are
impurities and integrates early-eluting peaks as described above.
[0138] Results
[0139] Two lots of Cystagon were dispensed in standard pharmacy containers
and
verified to be well within the manufacturer's expiration date. One lot was
provided by a
healthcare provider. It was dispensed in a standard pharmacy bottle and
verified by the
healthcare provider to be well within the expiration date. Upon analysis by
the Test Method,
it was shown to contain 9.1% cystamine by weight and 10.3% total related
substances, based
on the weight of cysteamine, using the assay described above. The second
analyzed
Cystagon lot was identified by lot number. Upon analysis by the assay
described above, it
was shown to contain 5.2% cystamine by weight and 5.7% total related
substances, based on
the weight of cysteamine. Each Cystagon lot was shipped and stored under
specified label
conditions.
[0140] Two representative lots of the CBGB capsule formulation were analyzed
by the
assay described above and were shown to contain 3.7% cystamine by weight and
3.6%
cystamine by weight, respectively, based on the weight of cysteamine, at the
time of
manufacture. For both lots, the total amount of related substances was 4.2% by
weight, based
on the weight of cysteamine.
[0141] The CBGB product lots were put on stability testing in various packages
and
storage conditions, then assayed for purity using the assay described above.
The results are
shown in the table below
-32-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
Lot Product Conditions Cystamine % / total related substances at time
point (month)
dose / count / C / % Initial 1 2 3 6 9 12
bottle size RH
1 75 mg / 60 / 100 25 / 60 3.7 / 3.7 / 3.1 / 3.4 /
3.5 / 3.7 / 3.8 /
cc 4.2 NA NA 4.2 4.4 5.1 5.5
1 75 mg / 60 / 100 40 / 75 3.7/ 3.7/ 3.2/ 3.5/ 3.9/
cc 4.2 NA NA 7.9 12.3
1 75 mg / 150 / 250 25 / 60 3.7 / 3.5 / 3.4 / 3.7 /
3.6 / 3.6 / 3.7 /
cc 4.2 NA NA 4.5 4.3 4.9 5.4
1 75 mg / 150 / 250 40 / 75 3.7/ 3.4/ 3.4/ 3.7/ 3.8/
cc 4.2 NA NA 7.9 11.6
1 75 mg / 300 / 400 25 / 60 3.7/ 3.5/ 3.3/ 3.4/ 3.5/
3.7/ 3.8/
cc 4.2 NA NA 4.2 4.4 5.1 5.7
1 75 mg / 300 / 400 40 / 75 3.7 / 3.4 / 3.2 / 3.6 /
4.0 /
cc 4.3 NA NA 7.7 12.8
1 75 mg / 60 / bulk 25 / 40 3.7/ 3.4/ 3.4/ 3.2/ 3.3/
3.3/ 3.2/
4.2 NA NA NA 4.2 4.5 4.6
1 75 mg / 60 / bulk 40 / 75 3.7/ 3.4/ 3.2/ 3.3/ 2.9/
4.2 NA NA NA 9.1
2 75 mg / 150 / 250 25 / 60 3.6 / 3.1 / 3.3 / 3.0 /
3.3 /
cc 4.2 4.0 4.3 4.3 5.0
2 75 mg / 150 / 250 40 / 75 3.6/ 3.1/ 3.6/
cc 4.2 7.5 12.1
[0142] Additional CBGB product samples according to Example 1 were put on long
term
stability testing in various packages and storage conditions, then assayed for
purity using the
assay described above. Results are shown in the table below.
Lot Product Conditions Cystamine % / total related substances at time
point (month)
dose/count/bottle C / % RH Initial 1 2 3 6 9 12 15 18
24
size
3 25 mg/60 /50 25 / 60 3.0 3.3 3.1 3.2 3.6
4.0
3.2 / 3.3 / 4.6
cc 4.1 / / /
4.7 / / NA /
/NA
NA NA 4.2 4.9 5.5 6.8
3 25 mg / 60 / 50 40 / 75 3.2 / 2.9 3.0 3.0
3.7 /
cc 4.1 13.4
NA NA 7.8
4 75 mg / 150 / 25 / 60 3 2 // 3.2 3.4 3.4 3.5
4.1 4.2 /
250 cc 4Ø / / / 5=2 4.7 . / /
NA /
NA
NA NA 4.7 = 5.3 6.3 7.1
4 75 mg / 150 / 40 / 75 3.2 / 3.1 3.4 3.5
4.0 /
250 cc 4.0 13.8
NA NA 9.0
-33-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
Lot Product Conditions Cystamine % / total related substances at time
point (month)
dose/count/bottle C / % RH Initial 1 2 3 6 9 12 15 18
24
size
75 mg / 60 / 100 25 / 60 3.4 3.5 3.3 3.5 5.0
3.9 3.9
3.4 / 3.7 / 5.3
/
cc 4.2 / / /
5.2 / / / /
NA NA 4.4 5.2 NA 6.0 NA NA
5 75 mg / 60 / 100 40 / 75 3.4/ 3.3 3.4 3.3
4.1/
/ / /
cc 4.2 16.0'
NA NA 8.5
5 75 mg / 300 / 25 / 60 3.5 3.5 3.5 3.3 5.3 4.1
4.
3.4 / 4.0 /
15.3 /
400 cc 4.2 / / /
6.0 / / / /
NA NA 4.9 5.3 NA 6.5 NA NA
5 75 mg / 300 / 40 / 75 3.4 / 3.5 3.7 3.7
4.2 /
/ / /
400 cc 4.2 15.4'
NA NA 9.6
5 75 mg / 60 / bulk 25 / 60 3.4 / 3.5 3.5 3.4
3.7 / 3.2 4.1
42 52 / / / / /
. .
NA NA NA = NA 5.5
5 75 mg / 60 / bulk 40 / 75 3.4 / 3.4 3.4 3.1 30/
/ / /
4.2 11.2
NA NA NA
6 25 mg / 60 / 50 25 / 60 3.3 3.2 3.2 3.3
4.0 3.9 4.4
3.3 / 3.7 / 5.1
/
cc 4.1 / / /
5.5 / / / /
NA NA 4.3 5.0 NA 5.9 NA NA
6 25 mg/60 /50 40 / 75 3.2 3.1 3.0
3.3 / 3.8/
cc 4.1 / / /
15.7'
NA NA 7.8
6 25 mg / 420 / 25 / 60 3.3 3.5 3.6 3.8 4.6 4.8
4.1
3.3 / 4.0 /
5.2/
250 cc 4.1 / / /
6.0 / / / /
NA NA 4.9 5.9 NA 7.4 NA NA
6 25 mg / 420 / 40 / 75 3.4 3.5 3.5
3.3 / 47/
250 cc 4.1 / / /
17.1'
NA NA 9.7
6 25 mg / 60 / bulk 25 / 60 3.4 3.4 3.3 3.1 3.5
3.3 / 3.7 /
41 54 / / / / /
. .
NA NA NA = NA 5.3
6 25 mg / 60 / bulk 40 / 75 3.3 / 3.3 3.2 2.9 2.9 /
/ / /
4.1 11.4
NA NA NA
7 75 mg / 60 / 100 25 / 60 3.2 3.3 3.3 3.2
4.6 4.1 3.6
3.2 / 3.5 / 4.6
/
cc 3.9 / / /
5.3 / / / /
NA NA 4.5 4.9 NA 6.1 NA NA
7 75 mg / 60 / 100 40 / 75 3.2/ 3.2 3.1 3.2 3.7/
3.9 / / / 13.5'
cc
NA NA 8.0
7 75 mg / 300 / 25 / 60 3.2 / 3.4 3.4 3.4 3.7 / 3.4
4.9 4.2 3.8 4.7 /
400 cc 3.9 / / / 5.5 / / / /
NA
NA NA 4.8 5.2 NA 6.4 NA
1
Samples pulled at 6 months but held at room temperature until new reference
standard was
qualified (at 8 months)
-34-

CA 02914770 2015-12-07
WO 2014/204881 PCT/US2014/042607
Lot Product Conditions Cystamine % / total related substances at time
point (month)
dose/count/bottle C / % RH Initial 1 2 3 6 9 12 15 18
24
size
7 75 mg / 300 / 40 / 75 3.2 / 3.3 3.3 3.5 3.9 /
400 cc 3.9 / / / 13.6'
NA NA 9.1
7 75 mg / 60 / bulk 25 / 60 3.2 / /
3.3 3.3 3.2 2.9 4.0
3.5
39 52 / / / / /
. .
NA NA NA = NA 5.5
7 75 mg / 60 / bulk 40 / 75 3.2 / 3.2 3.2 2.9
2.8 /
/ / /
3.9 10.3
NA NA NA
8 25 mg / 60 / 50 25 / 60 3.1 3.1 3.0 3.1 3.7
3.4 3.4
3.1 / 3.4 / 4.3 /
cc 3.9 / / /
4.9 / /
NA NA 4.1 4.9 NA 5.3 NA NA
8 25 mg/ 60 /50 40 / 75 3.1/ 3.0 2.9 2.7
3.4 /
/ / /
cc 3.9 13.4'
NA NA 7.4
8 25 mg / 420 / 25 / 60 8 3.2 3.3 3.3 3.4 4.1
4.5 3.
3.1 / 3.6 / 4.6 /
250 cc 3.9 / / /
5.3 / /
NA NA 4.6 5.0 NA 6.8 NA NA
8 25 mg / 420 / 40 / 75 3.3 3.3 3.
3.1 / 24.0 /
250 cc 3.9 / / /
16.2'
NA NA 9.1
8 25 mg / 60 / bulk 25 / 60 3.1/ 3.3 3.2 3.1 3.0 3.4
3.3 /
39 47 / / / / /
. .
NA NA NA = NA 4.9
8 25 mg / 60 / bulk 40 / 75 3.1 / 3.2 3.0 2.8 2.7 /
/ / /
3.9 10.6
NA NA NA
9 25 mg / 60 / 50 25 / 60 3.6 / /
3.5 2.9 3.2 3.3 3.4
3.4
cc 4.2 / / /
4.4 / /
NA NA 4.0 = 4.6 5.0
9 25 mg/ 60 /50 40 / 75 3.6/ 3.4 2.7 3.0
3.4 /
cc 4.2 / / /
11.4
NA NA 6.8
9 25 mg / 420 / 25 / 60 3.6 / /
3.5 3.0 3.4 3.5 3.8
3.4
250 cc 4.2 / / /
4 / /
. 4
NA NA 4.3 = 5.0 5.8
9 25 mg / 420 / 40 / 75 3.5 3.0 3.5
3.6 / 3.8 /
250 cc 4.2 / / /
12.9
NA NA 7.9
9 25 mg / 60 / bulk 25 / 40 3.6 / /
3.5 3.0 3.3 3.1 3.1
3.3
42 42 / / / / /
. .
NA NA NA = 4.2 4.6
9 25 mg / 60 / bulk 40 / 75 3.6 / 3.3 2.8 3.0
2.8 /
/ / /
4.2 9.1
NA NA NA
25 mg / 60 / 50 25 / 60 3.1 3.1/ 2.9 3.1
3.4 /
cc 4.0 NA NA /
4.1 / /
3.9 = 4.2 4.7
-35-

CA 02914770 2016-03-29
WO 2014/204881 PCT/US2014/042607
Lot Product Conditions Cystamine % / total related substances at time
point (month)
- -
dose/count/bottle C / % RH Initial 1 2 3 6 9 12 15 18 24
size
25 mg / 60 / 50 40 / 75 2.7 3.4 / 3.2
NA NA /
cc 4.0 .0 11.9
7
I Samples pulled at 6 months but held at room temperature until new reference
standard was qualified
(at 8 months)
[0143] All of the foregoing CBGB samples met the acid resistance criteria (Not
more than
10% (Q) of the label claim of cysteamine is dissolved after 2 hours in 0.1N
HC1) and
dissolution criteria (Not less than 70% (Q) of the label claim of cysteamine
is dissolved after
30 minutes in 0.2M sodium phosphate buffer, pH 6.8)
[0144] The foregoing description is given for clearness of understanding only,
and no
unnecessary limitations should be understood therefrom, as modifications
within the scope of
the invention may be apparent to those having ordinary skill in the art.
[0145] Throughout this specification and the claims which follow, unless the
context
requires otherwise, the word "comprise" and variations such as "comprises" and

"comprising" will be understood to imply the inclusion of a stated integer or
step or group of
integers or steps but not the exclusion of any other integer or step or group
of integers or
steps.
[0146] Throughout the specification, where compositions are described as
including
components or materials, it is contemplated that the compositions can also
consist essentially
of, or consist of, any combination of the recited components or materials,
unless described
otherwise. Likewise, where methods are described as including particular
steps, it is
contemplated that the methods can also consist essentially of, or consist of,
any combination
of the recited steps, unless described otherwise. The invention illustratively
disclosed herein
suitably may be practiced in the absence of any element or step which is not
specifically
disclosed herein.
[0147] The practice of a method disclosed herein, and individual steps
thereof, can be
performed manually and/or with the aid of or automation provided by electronic
equipment.
Although processes have been described with reference to particular
embodiments, a person
of ordinary skill in the art will readily appreciate that other ways of
performing the acts
associated with the methods may be used. For example, the order of various of
the steps may
-36-

CA 02914770 2016-03-29
W02014/204881
PCT/US2014/042607
be changed, unless described otherwise. In addition, some of the individual
steps can be
combined, omitted, or further subdivided into additional steps.
-37-

Representative Drawing

Sorry, the representative drawing for patent document number 2914770 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2016-09-27
(86) PCT Filing Date 2014-06-17
(87) PCT Publication Date 2014-12-24
(85) National Entry 2015-12-07
Examination Requested 2016-01-05
(45) Issued 2016-09-27

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $347.00 was received on 2024-06-07


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2025-06-17 $347.00 if received in 2024
$362.27 if received in 2025
Next Payment if small entity fee 2025-06-17 $125.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2015-12-07
Advance an application for a patent out of its routine order $500.00 2016-01-05
Request for Examination $800.00 2016-01-05
Maintenance Fee - Application - New Act 2 2016-06-17 $100.00 2016-05-25
Final Fee $300.00 2016-08-04
Registration of a document - section 124 $100.00 2017-06-01
Maintenance Fee - Patent - New Act 3 2017-06-19 $100.00 2017-06-12
Maintenance Fee - Patent - New Act 4 2018-06-18 $300.00 2018-06-25
Maintenance Fee - Patent - New Act 5 2019-06-17 $400.00 2019-07-05
Maintenance Fee - Patent - New Act 6 2020-06-17 $200.00 2020-06-12
Maintenance Fee - Patent - New Act 7 2021-06-17 $204.00 2021-06-11
Maintenance Fee - Patent - New Act 8 2022-06-17 $203.59 2022-06-10
Maintenance Fee - Patent - New Act 9 2023-06-19 $210.51 2023-06-12
Registration of a document - section 124 2023-10-24 $100.00 2023-10-24
Maintenance Fee - Patent - New Act 10 2024-06-17 $347.00 2024-06-07
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
HORIZON THERAPEUTICS U.S. HOLDING LLC
Past Owners on Record
HORIZON ORPHAN LLC
RAPTOR PHARMACEUTICALS INC.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Claims 2015-12-08 7 295
Abstract 2015-12-07 1 45
Claims 2015-12-07 7 289
Description 2015-12-07 37 1,972
Cover Page 2016-02-04 1 26
Claims 2016-03-29 8 332
Description 2016-03-29 37 1,964
Claims 2016-05-24 5 197
Cover Page 2016-08-31 1 26
Prosecution-Amendment 2016-01-11 1 24
Patent Cooperation Treaty (PCT) 2015-12-07 1 39
International Search Report 2015-12-07 2 97
National Entry Request 2015-12-07 5 135
Voluntary Amendment 2015-12-07 2 45
Special Order 2016-01-05 2 69
Examiner Requisition 2016-02-01 5 309
Amendment 2016-03-29 27 1,112
Examiner Requisition 2016-05-02 5 310
Amendment 2016-05-24 8 354
Final Fee 2016-08-04 2 47
Maintenance Fee Payment 2023-06-12 3 53